1
|
Wang L, Huan XJ, Song SS, Bao XB, Tian CQ, Miao ZH, Wang YQ. UBE4B modulates BET inhibitor sensitivity via KLHL22-JAK2-PIM1 axis in hepatocellular carcinoma. Biochem Pharmacol 2025; 237:116943. [PMID: 40228637 DOI: 10.1016/j.bcp.2025.116943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/18/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Ubiquitination factor E4B (UBE4B) is crucial to the high mortality rate and poor prognosis associated with hepatocellular carcinoma (HCC). Evidence suggests that aberrant epigenetic modifications significantly contribute to HCC carcinogenesis, making epigenetic mechanisms a promising area for therapeutic intervention. However, the precise role of UBE4B in the epigenetic dysregulation observed in HCC remains elusive. In this study, we silenced UBE4B in HCC cells and exposed them to a panel of epigenetic compounds. Notably, only bromodomain and extraterminal inhibitors (BETis) exhibited resistance to UBE4B silencing, while restoring UBE4B expression partially reversed this resistance. Furthermore, UBE4B deletion led to decreased growth rates and impaired proliferation, resulting in cell cycle arrest and diminished tumorigenicity. However, this deletion did not affect the cell cycle arrest induced by BETi. Interestingly, KLHL22, a ubiquitin substrate of UBE4B, accumulated in UBE4B-deleted cells. Knockdown of KLHL22 restored sensitivity to BETi, accompanied by downregulation of JAK2 and upregulation of its negative regulator, LNK. Additionally, UBE4B deletion resulted in decreased LNK expression, and LNK knockdown increased JAK2 expression and mediated resistance to BETi. Increased JAK2 subsequently targeted PIM1, further reducing the inhibitory effect of BETi. Directly silencing PIM1 in UBE4B-deleted cells restored BETi sensitivity. Overall, our findings provide novel insights into the relationship between UBE4B expression and BETi sensitivity, which is mediated through the KLHL22-JAK2-PIM1 regulatory axis. These findings not only deepen our understanding of the mechanisms underlying HCC progression but also suggest that targeting this axis may present a promising therapeutic strategy for enhancing the treatment outcomes of HCC.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xia-Juan Huan
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | - Shan-Shan Song
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | - Xu-Bin Bao
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | - Chang-Qing Tian
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | - Ze-Hong Miao
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Ying-Qing Wang
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|
2
|
Han L, Sun Y, Yang K, Long C. Inhibition of JAK2/STAT3 by pacritinib synergizes with chemotherapy in esophageal carcinoma. Toxicol In Vitro 2025; 106:106056. [PMID: 40112935 DOI: 10.1016/j.tiv.2025.106056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
The poor outcomes associated with esophageal carcinoma, particularly in advanced stages, necessitate the development of new treatment strategies. This study examines the efficacy of pacritinib, a multi-kinase inhibitor, both alone and in combination with carboplatin, in preclinical esophageal carcinoma models. Six esophageal carcinoma cell lines (KYSE-70, OE33, FLO-1, KYAE-1, ESO 26, and HCE-6) were treated with pacritinib, resulting in a dose-dependent reduction in cell viability. Combination index (CI) analysis demonstrated strong synergy between pacritinib and carboplatin across this panel of cell lines. In in vivo esophageal carcinoma xenograft model, pacritinib alone significantly reduced tumor growth and improved survival rates compared to control. Notably, the combination of pacritinib and carboplatin further reduced tumor growth and improved survival rates compared to either treatment alone. Toxicity assessment showed that neither single-agent nor combination treatment resulted in significantly altered levels of body weight and serum markers, supporting the safety profile of pacritinib in combination with carboplatin. Mechanistic studies revealed that while pacritinib inhibited the phosphorylation of JAK, STAT3, and IRAK1 in esophageal carcinoma cells, it is the suppression of the JAK/STAT3 pathway, rather than IRAK1, that is responsible for the synergistic effect with carboplatin. Our findings indicate that pacritinib possesses potent anti-tumor activity in esophageal carcinoma and enhances the efficacy of carboplatin through the suppression of JAK/STAT3 signaling, warranting further clinical investigation.
Collapse
Affiliation(s)
- Lan Han
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Yinyin Sun
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Kai Yang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China.
| | - Cheng Long
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China.
| |
Collapse
|
3
|
Roskoski R. Properties of FDA-approved small molecule protein kinase inhibitors: A 2025 update. Pharmacol Res 2025; 216:107723. [PMID: 40252783 DOI: 10.1016/j.phrs.2025.107723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/21/2025]
Abstract
Because of the deregulation of protein kinase action in many inflammatory diseases and cancer, the protein kinase family has become one of the most significant drug targets in the 21st century. There are 85 FDA-approved protein kinase antagonists that target about two dozen different enzymes and four of these drugs were approved in 2024 and a fifth was approved in 2025. Of these drugs, five target dual specificity protein kinases (MEK1/2), fourteen inhibit protein-serine/threonine protein kinases, twenty-one block nonreceptor protein-tyrosine kinases, and 45 target receptor protein-tyrosine kinases. The data indicate that 75 of these drugs are prescribed for the treatment of neoplasms. Seven drugs (abrocitinib, baricitinib, deucravacitinib, deuruxolitinib, ritlecitinib, tofacitinib, upadacitinib) are prescribed for the management of inflammatory diseases (atopic dermatitis, rheumatoid arthritis, psoriasis, alopecia areata, and ulcerative colitis). Of the 85 FDA-approved agents, about two dozen are used in the treatment of multiple diseases. The following four drugs received FDA approval in 2024 - deuruxolitinib (alopecia areata), ensartinib and lazertinib (non-small cell lung cancer), and tovorafenib (pediatric glioma) while mirdametinib was approved in 2025 for the treatment of type I neurofibromatosis (von Recklinghausen disease). Apart from netarsudil, temsirolimus, and trilaciclib, the approved protein kinase blockers are orally bioavailable. This article summarizes the physicochemical properties of all 85 FDA-approved small molecule protein kinase inhibitors including the molecular weight, number of hydrogen bond donors/acceptors, ligand efficiency, lipophilic efficiency, polar surface area, and solubility. A total of 39 of the 85 FDA-approved drugs have a least one Lipinski rule of 5 violation.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 221 Haywood Knolls Drive, Hendersonville, NC 28791, United States.
| |
Collapse
|
4
|
Ghosh S, Finnemann SC, Vollrath D, Rothlin CV. In the Eyes of the Beholder-New Mertk Knockout Mouse and Re-Evaluation of Phagocytosis versus Anti-Inflammatory Functions of MERTK. Int J Mol Sci 2024; 25:5299. [PMID: 38791338 PMCID: PMC11121519 DOI: 10.3390/ijms25105299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Greg Lemke's laboratory was one of the pioneers of research into the TAM family of receptor tyrosine kinases (RTKs). Not only was Tyro3 cloned in his laboratory, but his group also extensively studied mice knocked out for individual or various combinations of the TAM RTKs Tyro3, Axl, and Mertk. Here we primarily focus on one of the paralogs-MERTK. We provide a historical perspective on rodent models of loss of Mertk function and their association with retinal degeneration and blindness. We describe later studies employing mouse genetics and the generation of newer knockout models that point out incongruencies with the inference that loss of MERTK-dependent phagocytosis is sufficient for severe, early-onset photoreceptor degeneration in mice. This discussion is meant to raise awareness with regards to the limitations of the original Mertk knockout mouse model generated using 129 derived embryonic stem cells and carrying 129 derived alleles and the role of these alleles in modifying Mertk knockout phenotypes or even displaying Mertk-independent phenotypes. We also suggest molecular approaches that can further Greg Lemke's scintillating legacy of dissecting the molecular functions of MERTK-a protein that has been described to function in phagocytosis as well as in the negative regulation of inflammation.
Collapse
Affiliation(s)
- Sourav Ghosh
- Department of Neurology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA;
| | - Douglas Vollrath
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Carla V. Rothlin
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
5
|
Mao F, Gao L, Liu L, Tang Y. Enhanced synergy of pacritinib with temsirolimus and sunitinib in preclinical renal cell carcinoma model by targeting JAK2/STAT pathway. J Chemother 2024; 36:238-248. [PMID: 37916436 DOI: 10.1080/1120009x.2023.2274700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023]
Abstract
Pacritinib is an oral medication that inhibits several kinases including JAK2, FLT3, IRAK and STAT3. It has been recently approved to treat patients with thrombocytopenia and myelofibrosis. Studies are currently exploring the potential use of pacritinib in treating other types of cancer such as leukaemia, breast cancer and prostate cancer. Our study aimed to investigate the effects of pacritinib alone and its combination with standard of care in renal cell carcinoma (RCC). We showed that pacritinib dose-dependently decreased viability of RCC cells, with IC50 at nanomolar or low micromolar concentration rage. Pacritinib inhibited cell proliferation, decreased colony formation, and increased apoptosis. Interestingly, pacritinib exhibited synergistic effects when combined with temsirolimus and sunitinib, but antagonistic effects when combined with doxorubicin, in a panel of RCC cell lines. We also confirmed that the combination of pacritinib with temsirolimus and sunitinib resulted in synergistic effects in RCC mouse models, with complete inhibition of tumour growth throughout the treatment period. Mechanistic studies indicated that the inhibition of JAK2, but not IRAK, was the main contributor to the anti-RCC activity of pacritinib. Our study is the first to demonstrate that pacritinib shows promise as a treatment option for RCC and underscores the therapeutic potential of targeting the JAK2/STAT signalling pathway in RCC.
Collapse
Affiliation(s)
- Fei Mao
- Department of Urology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| | - Liangkui Gao
- Department of Urology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| | - Liming Liu
- Department of Urology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| | - Yuanjia Tang
- Department of Urology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| |
Collapse
|
6
|
Roskoski R. Cost in the United States of FDA-approved small molecule protein kinase inhibitors used in the treatment of neoplastic and non-neoplastic diseases. Pharmacol Res 2024; 199:107036. [PMID: 38096958 DOI: 10.1016/j.phrs.2023.107036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023]
Abstract
Because genetic alterations including mutations, overexpression, translocations, and dysregulation of protein kinases are involved in the pathogenesis of many illnesses, this enzyme family is the target of many drug discovery programs worldwide. The FDA has approved 80 small molecule protein kinase inhibitors with 77 drugs orally bioavailable. The data indicate that 69 of these medicinals are approved for the management of neoplasms including solid tumors such as breast and lung cancer as well as non-solid tumors such as leukemia. Moreover, the remaining 11 drugs target non-neoplastic diseases including psoriasis, rheumatoid arthritis, and ulcerative colitis. The cost of drugs was obtained from www.pharmacychecker.com using the FDA label to determine the dosage and number of tablets required per day. This methodology excludes any private or governmental insurance coverage, which would cover the entire cost or more likely a fraction of the stated price. The average monthly cost for the treatment of neoplastic diseases was $17,900 with a price of $44,000 for futibatinib (used to treat cholangiocarcinomas with FGFR2 fusions) and minimum of $5100 for binimetinib (melanoma). The average monthly cost for the treatment of non-neoplastic diseases was $6800 with a maximum of $17,000 for belumosudil (graft vs. host disease) and a minimum of $200 for netarsudil eye drops (glaucoma). There is a negative correlation of the cost of the drugs and the incidence of the targeted disease. Many of these agents are or were designated as orphan drugs meaning that there are fewer than 200,000 potential patients in the United States.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 221 Haywood Knolls Drive, Hendersonville, NC 28791, United States.
| |
Collapse
|
7
|
Sarapultsev A, Gusev E, Komelkova M, Utepova I, Luo S, Hu D. JAK-STAT signaling in inflammation and stress-related diseases: implications for therapeutic interventions. MOLECULAR BIOMEDICINE 2023; 4:40. [PMID: 37938494 PMCID: PMC10632324 DOI: 10.1186/s43556-023-00151-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
The Janus kinase-signal transducer and transcription activator pathway (JAK-STAT) serves as a cornerstone in cellular signaling, regulating physiological and pathological processes such as inflammation and stress. Dysregulation in this pathway can lead to severe immunodeficiencies and malignancies, and its role extends to neurotransduction and pro-inflammatory signaling mechanisms. Although JAK inhibitors (Jakinibs) have successfully treated immunological and inflammatory disorders, their application has generally been limited to diseases with similar pathogenic features. Despite the modest expression of JAK-STAT in the CNS, it is crucial for functions in the cortex, hippocampus, and cerebellum, making it relevant in conditions like Parkinson's disease and other neuroinflammatory disorders. Furthermore, the influence of the pathway on serotonin receptors and phospholipase C has implications for stress and mood disorders. This review expands the understanding of JAK-STAT, moving beyond traditional immunological contexts to explore its role in stress-related disorders and CNS function. Recent findings, such as the effectiveness of Jakinibs in chronic conditions such as rheumatoid arthritis, expand their therapeutic applicability. Advances in isoform-specific inhibitors, including filgotinib and upadacitinib, promise greater specificity with fewer off-target effects. Combination therapies, involving Jakinibs and monoclonal antibodies, aiming to enhance therapeutic specificity and efficacy also give great hope. Overall, this review bridges the gap between basic science and clinical application, elucidating the complex influence of the JAK-STAT pathway on human health and guiding future interventions.
Collapse
Affiliation(s)
- Alexey Sarapultsev
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080, Chelyabinsk, Russia.
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia.
| | - Evgenii Gusev
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080, Chelyabinsk, Russia
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia
| | - Maria Komelkova
- Russian-Chinese Education and Research Center of System Pathology, South Ural State University, 454080, Chelyabinsk, Russia
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia
| | - Irina Utepova
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 620002, Ekaterinburg, Russian Federation
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, 430022, China
- Clinical Research Center of Cancer Immunotherapy, Hubei Wuhan, 430022, China
| |
Collapse
|
8
|
Yunianto I, Currie M, Chitcholtan K, Sykes P. Potential drug repurposing of ruxolitinib to inhibit the JAK/STAT pathway for the treatment of patients with epithelial ovarian cancer. J Obstet Gynaecol Res 2023; 49:2563-2574. [PMID: 37565583 DOI: 10.1111/jog.15761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
AIM This review aimed to describe the potential for therapeutic targeting of the JAK/STAT signaling pathway by repurposing the clinically-approved JAK inhibitor ruxolitinib in the patients with epithelial ovarian cancer (OC) setting. METHODS We reviewed publications that focus on the inhibition of the JAK/STAT pathway in hematological and solid malignancies including OC. RESULTS Preclinical studies showed that ruxolitinib effectively reduces OC cell viability and metastasis and enhances the anti-tumor activity of chemotherapy drugs. There are a number of recent clinical trials exploring the role of JAK/STAT inhibition in solid cancers including OC. Early results have not adequately supported efficacy in solid tumors. However, there are preclinical data and clinical studies supporting the use of ruxolitinib in combination with both chemotherapy and other targeted drugs in OC setting. CONCLUSION Inflammatory conditions and persistent activation of the JAK/STAT pathway are associated with tumourigenesis and chemoresistance, and therapeutic blockade of this pathway shows promising results. For women with OC, clinical investigation exploring the role of ruxolitinib in combination with chemotherapy agents or other targeted therapeutics is warranted.
Collapse
Affiliation(s)
- Irfan Yunianto
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
- Department of Biology Education, Universitas Ahmad Dahlan, Indonesia
| | - Margaret Currie
- Department of Pathology and Biomedical Sciences, University of Otago, Christchurch, New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
9
|
Roskoski R. Deucravacitinib is an allosteric TYK2 protein kinase inhibitor FDA-approved for the treatment of psoriasis. Pharmacol Res 2023; 189:106642. [PMID: 36754102 DOI: 10.1016/j.phrs.2022.106642] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 12/31/2022] [Indexed: 02/09/2023]
Abstract
Psoriasis is a heterogeneous, inflammatory, autoimmune skin disease that affects up to 2% of the world's population. There are many treatment modalities including topical medicines, ultraviolet light therapy, monoclonal antibodies, and several oral medications. Cytokines play a central role in the pathogenesis of this disorder including TNF-α, (tumor necrosis factor-α) IL-17A (interleukin-17A), IL-17F, IL-22, and IL-23. Cytokine signaling involves transduction mediated by the JAK-STAT pathway. There are four JAKS (JAK1/2/3 and TYK2) and six STATS (signal transducer and activators of transcription). Janus kinases contain an inactive JH2 domain that is aminoterminal to the active JH1 domain. Under basal conditions, the JH2 domain inhibits the activity of the JH1 domain. Deucravacitinib is an orally effective N-trideuteromethyl-pyridazine derivative that targets and stabilizes the TYK2 JH2 domain and thereby blocks TYK2 JH1 activity. Seven other JAK inhibitors, which target the JAK family JH1 domain, are prescribed for the treatment of neoplastic and other inflammatory diseases. The use of deuterium in the trimethylamide decreases the rate of demethylation and slows the production of a metabolite that is active against a variety of targets in addition to TYK2. A second unique aspect in the development of deucravacitinib is the targeting of a pseudokinase domain. Deucravacitinib is rather specific for TYK2 and its toxic effects are much less than those of the other FDA-approved JAK inhibitors. The successful development of deucravacitinib may stimulate the development of additional pseudokinase ligands for the JAK family and for other kinase families as well.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 106, Box 19, Horse Shoe, NC 28742-8814, United States.
| |
Collapse
|
10
|
Sharma V, Gupta M. Designing of kinase hinge binders: A medicinal chemistry perspective. Chem Biol Drug Des 2022; 100:968-980. [PMID: 35112799 DOI: 10.1111/cbdd.14024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 01/16/2022] [Accepted: 01/29/2022] [Indexed: 01/25/2023]
Abstract
Protein kinases are key regulators of cellular signaling and play a critical role in oncogenesis. Inhibitors of protein kinases are pursued by both industry and academia as a promising target for cancer therapy. Within the protein kinases, the ATP site has produced more than 40 FDA-approved drugs. The ATP site is broadly composed of a hinge region, gatekeeper residues, DFG-loop, ribose pocket, and other hydrophobic regions. The hinge region in the ATP site can be used for designing potent inhibitors. In this review, we discuss some representative studies that will highlight the interactions of heterocyclic compounds with hinge regions of different kinases like BRAF kinase, EGRF kinase, MAP kinase, and Mps1 kinase.
Collapse
Affiliation(s)
- Vikas Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Mohit Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon, USA.,GreenLight Biosciences, Woburn, MA, United States
| |
Collapse
|
11
|
Zhang HG, Wang B, Yang Y, Liu X, Wang J, Xin N, Li S, Miao Y, Wu Q, Guo T, Yuan Y, Zuo Y, Chen X, Ren T, Dong C, Wang J, Ruan H, Sun M, Xu X, Zheng H. Depression compromises antiviral innate immunity via the AVP-AHI1-Tyk2 axis. Cell Res 2022; 32:897-913. [PMID: 35821088 PMCID: PMC9274186 DOI: 10.1038/s41422-022-00689-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/24/2022] [Indexed: 12/13/2022] Open
Abstract
Depression is a serious public-health issue. Recent reports have suggested higher susceptibility to viral infections in depressive patients. However, how depression affects antiviral innate immune signaling remains unknown. Here, we revealed a reduction in expression of Abelson helper integration site 1 (AHI1) in the peripheral blood mononuclear cells (PBMCs) and macrophages from the patients with major depressive disorder (MDD), which leads to attenuated antiviral immune response. We found that depression-related arginine vasopressin (AVP) induces reduction of AHI1 in macrophages. Further studies demonstrated that AHI1 is a critical stabilizer of basal type-I-interferon (IFN-I) signaling. Mechanistically, AHI1 recruits OTUD1 to deubiquitinate and stabilize Tyk2, while AHI1 reduction downregulates Tyk2 and IFN-I signaling activity in macrophages from both MDD patients and depression model mice. Interestingly, we identified a clinical analgesic meptazinol that effectively stimulates AHI1 expression, thus enhancing IFN-I antiviral defense in depression model mice. Our study promotes the understanding of the signaling mechanisms of depression-mediated antiviral immune dysfunction, and reveals meptazinol as an enhancer of antiviral innate immunity in depressive patients.
Collapse
Affiliation(s)
- Hong-Guang Zhang
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Bin Wang
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yong Yang
- Department of Psychiatry, the Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuan Liu
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Junjie Wang
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Ning Xin
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Shifeng Li
- Department of Intensive Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ying Miao
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Qiuyu Wu
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Tingting Guo
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yukang Yuan
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Yibo Zuo
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Xiangjie Chen
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Tengfei Ren
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Chunsheng Dong
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China
| | - Jun Wang
- Department of Intensive Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hang Ruan
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xingshun Xu
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China.
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China.
| | - Hui Zheng
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China.
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
12
|
Kumar V, Lee G, Yoo J, Ro HS, Lee KW. An attention mechanism-based LSTM network for cancer kinase activity prediction. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2022; 33:631-647. [PMID: 36062308 DOI: 10.1080/1062936x.2022.2109062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/30/2022] [Indexed: 06/15/2023]
Abstract
Despite the endeavours and achievements made in treating cancers during the past decades, resistance to available kinase drugs continues to be a major problem in cancer therapies. Thus, it is highly desirable to develop computational models that can predict the bioactivity of a compound against cancer kinases. Here, we present a Long Short-Term Memory (LSTM) framework for predicting the activities of lead molecules against seven different kinases. A total of 14,907 compounds from the ChEMBL database were selected for model building. Two different molecular representations, namely, 2D descriptors and MACCS fingerprints were subjected to the LSTM method for the training process. We also successfully integrated an attention mechanism into our model, which helped us to interpret the contribution of chemical features on kinase activity. The attention mechanism extracted the significant chemical moieties more effectively by taking them into consideration during the activity prediction. The recorded accuracies in the test sets for both 2D descriptors and MACCS fingerprints-based models were 0.81 and 0.78, respectively. The receiver operating characteristic curve (ROC)-area under the curve (AUC) score for both models was in the range of 0.8-0.99. The proposed framework can be a good starting point for the development of new cancer kinase drugs.
Collapse
Affiliation(s)
- V Kumar
- Department of Bio & Medical Big Data (BK21 Four Program), Division of Life Sciences, Research Institute of Life Sciences, Gyeongsang National University, Jinju, Korea
| | - G Lee
- Division of Applied Life Science (BK21 Program), ABC-RLRC, PMBBRC, Gyeongsang National University, Jinju, Korea
| | - J Yoo
- Division of Applied Life Science (BK21 Program), Research Institute of Life Sciences, Gyeongsang National University, Jinju, Korea
| | - H S Ro
- Department of Bio & Medical Big Data (BK21 Four Program), Division of Life Sciences, Research Institute of Life Sciences, Gyeongsang National University, Jinju, Korea
| | - K W Lee
- Department of Bio & Medical Big Data (BK21 Four Program), Division of Life Sciences, Research Institute of Life Sciences, Gyeongsang National University, Jinju, Korea
- ANGEL i-Drug Design (AiDD), Jinju, Korea
| |
Collapse
|
13
|
Roskoski R. Janus kinase (JAK) inhibitors in the treatment of neoplastic and inflammatory disorders. Pharmacol Res 2022; 183:106362. [PMID: 35878738 DOI: 10.1016/j.phrs.2022.106362] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 02/07/2023]
Abstract
The Janus kinase (JAK) family of nonreceptor protein-tyrosine kinases consists of JAK1, JAK2, JAK3, and TYK2 (Tyrosine Kinase 2). Each of these proteins contains a JAK homology pseudokinase (JH2) domain that interacts with and regulates the activity of the adjacent protein kinase domain (JH1). The Janus kinase family is regulated by numerous cytokines including interferons, interleukins, and hormones such as erythropoietin and thrombopoietin. Ligand binding to cytokine receptors leads to the activation of associated Janus kinases, which then catalyze the phosphorylation of the receptors. The SH2 domain of signal transducers and activators of transcription (STAT) binds to the cytokine receptor phosphotyrosines thereby promoting STAT phosphorylation and activation by the Janus kinases. STAT dimers are then translocated into the nucleus where they participate in the regulation and expression of dozens of proteins. JAK1/3 signaling participates in the pathogenesis of inflammatory disorders while JAK1/2 signaling contributes to the development of myeloproliferative neoplasms as well as several malignancies including leukemias and lymphomas. An activating JAK2 V617F mutation occurs in 95% of people with polycythemia vera and about 50% of cases of myelofibrosis and essential thrombocythemia. Abrocitinib, ruxolitinib, and upadacitinib are JAK inhibitors that are FDA-approved for the treatment of atopic dermatitis. Baricitinib is used for the treatment of rheumatoid arthritis and covid 19. Tofacitinib and upadacitinib are JAK antagonists that are used for the treatment of rheumatoid arthritis and ulcerative colitis. Additionally, ruxolitinib is approved for the treatment of polycythemia vera while fedratinib, pacritinib, and ruxolitinib are approved for the treatment of myelofibrosis.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 106, Box 19, Horse Shoe, NC 28742, United States.
| |
Collapse
|
14
|
Early Use of Low-Dose Ruxolitinib: A Promising Strategy for the Treatment of Acute and Chronic GVHD. Pharmaceuticals (Basel) 2022; 15:ph15030374. [PMID: 35337171 PMCID: PMC8955311 DOI: 10.3390/ph15030374] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/09/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023] Open
Abstract
Janus kinases (JAK) are a family of tyrosine kinases (JAK1, JAK2, JAK3, and TYK2) that transduce cytokine-mediated signals through the JAK–STAT metabolic pathway. These kinases act by regulating the transcription of specific genes capable of inducing biological responses in several immune cell subsets. Inhibition of Janus kinases interferes with the JAK–STAT signaling pathway. Besides being used in the treatment of cancer and inflammatory diseases, in recent years, they have also been used to treat inflammatory conditions, such as graft-versus-host disease (GVHD) and cytokine release syndrome as complications of allogeneic hematopoietic stem cell transplantation and cell therapy. Recently, the FDA approved the use of ruxolitinib, a JAK1/2 inhibitor, in the treatment of acute steroid-refractory GVHD (SR-aGVHD), highlighting the role of JAK inhibition in this immune deregulation. Ruxolitinib was initially used to treat myelofibrosis and true polycythemia in a high-dose treatment and caused hematological toxicity. Since a lower dosage often could not be effective, the use of ruxolitinib was suspended. Subsequently, ruxolitinib was evaluated in adult patients with SR-aGVHD and was found to achieve a rapid and effective response. In addition, its early low-dose use in pediatric patients affected by GVHD has proved effective, safe, and reasonably preventive. The review aims to describe the potential properties of ruxolitinib to identify new therapeutic strategies.
Collapse
|
15
|
A novel graph convolutional neural network for predicting interaction sites on protein kinase inhibitors in phosphorylation. Sci Rep 2022; 12:229. [PMID: 34997142 PMCID: PMC8742007 DOI: 10.1038/s41598-021-04230-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/14/2021] [Indexed: 11/08/2022] Open
Abstract
Protein kinase-inhibitor interactions are key to the phosphorylation of proteins involved in cell proliferation, differentiation, and apoptosis, which shows the importance of binding mechanism research and kinase inhibitor design. In this study, a novel machine learning module (i.e., the WL Box) was designed and assembled to the Prediction of Interaction Sites of Protein Kinase Inhibitors (PISPKI) model, which is a graph convolutional neural network (GCN) to predict the interaction sites of protein kinase inhibitors. The WL Box is a novel module based on the well-known Weisfeiler-Lehman algorithm, which assembles multiple switch weights to effectively compute graph features. The PISPKI model was evaluated by testing with shuffled datasets and ablation analysis using 11 kinase classes. The accuracy of the PISPKI model with the shuffled datasets varied from 83 to 86%, demonstrating superior performance compared to two baseline models. The effectiveness of the model was confirmed by testing with shuffled datasets. Furthermore, the performance of each component of the model was analyzed via the ablation study, which demonstrated that the WL Box module was critical. The code is available at https://github.com/feiqiwang/PISPKI .
Collapse
|
16
|
Hromadová D, Elewaut D, Inman RD, Strobl B, Gracey E. From Science to Success? Targeting Tyrosine Kinase 2 in Spondyloarthritis and Related Chronic Inflammatory Diseases. Front Genet 2021; 12:685280. [PMID: 34290741 PMCID: PMC8287328 DOI: 10.3389/fgene.2021.685280] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 12/16/2022] Open
Abstract
Spondyloarthritis (SpA) is a family of inflammatory arthritic diseases, which includes the prototypes of psoriatic arthritis and ankylosing spondylitis. SpA is commonly associated with systemic inflammatory diseases, such as psoriasis and inflammatory bowel disease. Immunological studies, murine models and the genetics of SpA all indicate a pathogenic role for the IL-23/IL-17 axis. Therapeutics targeting the IL-23/IL-17 pathway are successful at providing symptomatic relief, but may not provide complete protection against progression of arthritis. Thus there is still tremendous interest in the discovery of novel therapeutic targets for SpA. Tyrosine kinase 2 (TYK2) is a member of the Janus kinases, which mediate intracellular signaling of cytokines via signal transducer and activator of transcription (STAT) activation. TYK2 plays a crucial role in mediating IL-23 receptor signaling and STAT3 activation. A plethora of natural mutations in and around TYK2 have provided a wealth of data to associate this kinase with autoimmune/autoinflammatory diseases in humans. Induced and natural mutations in murine Tyk2 largely support human data; however, key inter-species differences exist, which means extrapolation of data from murine models to humans needs to be done with caution. Despite these reservations, novel selective TYK2 inhibitors are now proving successful in advanced clinical trials of inflammatory diseases. In this review, we will discuss TYK2 from basic biology to therapeutic targeting, with an emphasis on studies in SpA. Seminal studies uncovering the basic science of TYK2 have provided sound foundations for targeting it in SpA and related inflammatory diseases. TYK2 inhibitors may well be the next blockbuster therapeutic for SpA.
Collapse
Affiliation(s)
- Dominika Hromadová
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dirk Elewaut
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Robert D. Inman
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Departments of Medicine and Immunology, University of Toronto, Toronto, ON, Canada
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eric Gracey
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
17
|
Properties of FDA-approved small molecule protein kinase inhibitors: A 2021 update. Pharmacol Res 2021; 165:105463. [DOI: 10.1016/j.phrs.2021.105463] [Citation(s) in RCA: 249] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
|
18
|
Kim D, Park M, Haleem I, Lee Y, Koo J, Na YC, Song G, Lee J. Natural Product Ginsenoside 20(S)-25-Methoxyl-Dammarane-3β, 12β, 20-Triol in Cancer Treatment: A Review of the Pharmacological Mechanisms and Pharmacokinetics. Front Pharmacol 2020; 11:521. [PMID: 32425780 PMCID: PMC7212460 DOI: 10.3389/fphar.2020.00521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/02/2020] [Indexed: 01/01/2023] Open
Abstract
Panax ginseng has been used as an herbal medicine for thousands of years. Most of its pharmacological effects are attributed to its constituent ginsenosides, including 20(S)-25-methoxyl-dammarane-3β, 12β, 20-triol (20(S)-25-OCH3-PPD), which is one of the protopanaxadiol type ginsenosides. It has been found to exhibit anticancer effects by interacting with multiple pharmacological pathways, such as the Wnt/β-catenin, MDM2, ERK/MAPK, and STAT3 signaling pathways. However, its therapeutic potential could be limited by its low bioavailability mainly due to its low aqueous solubility. Thus, several studies have been conducted on its pharmacokinetics and its delivery systems, so as to increase its oral bioavailability. In this review, comprehensive information on its varying pharmacological pathways in cancer, as well as its pharmacokinetic behavior and pharmaceutical strategies, is provided. This information would be useful in the understanding of its diverse mechanisms and pharmacokinetics as an anticancer drug, leading to the design of superior 20(S)-25-OCH3-PPD-containing formulations that maximize its therapeutic potential.
Collapse
Affiliation(s)
- Dohyun Kim
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Minwoo Park
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Iqra Haleem
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Younghong Lee
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Jain Koo
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Young Chae Na
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Gidong Song
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| |
Collapse
|
19
|
Arshad S, Naveed M, Ullia M, Javed K, Butt A, Khawar M, Amjad F. Targeting STAT-3 signaling pathway in cancer for development of novel drugs: Advancements and challenges. Genet Mol Biol 2020; 43:e20180160. [PMID: 32167126 PMCID: PMC7198026 DOI: 10.1590/1678-4685-gmb-2018-0160] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Signal transducers and activators of transcription 3 (STAT-3) is a transcription
factor that regulates the gene expression of several target genes. These factors
are activated by the binding of cytokines and growth factors with STAT-3
specific receptors on cell membrane. Few years ago, STAT-3 was considered an
acute phase response element having several cellular functions such as
inflammation, cell survival, invasion, metastasis and proliferation, genetic
alteration, and angiogenesis. STAT-3 is activated by several types of
inflammatory cytokines, carcinogens, viruses, growth factors, and oncogenes.
Thus, the STAT3 pathway is a potential target for cancer therapeutics. Abnormal
STAT-3 activity in tumor development and cellular transformation can be targeted
by several genomic and pharmacological methodologies. An extensive review of the
literature has been conducted to emphasize the role of STAT-3 as a unique cancer
drug target. This review article discusses in detail the wide range of STAT-3
inhibitors that show antitumor effects both in vitro and
in vivo. Thus, targeting constitutive STAT-3 signaling is a
remarkable therapeutic methodology for tumor progression. Finally, current
limitations, trials and future perspectives of STAT-3 inhibitors are also
critically discussed.
Collapse
Affiliation(s)
- Sundas Arshad
- University of Lahore, Department of Allied Health Sciences, Gujrat Campus, Pakistan
| | - Muhammad Naveed
- University of Central Punjab, Faculty of life sciences, Department of Biotechnology, Lahore, Pakistan
| | - Mahad Ullia
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Khadija Javed
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Ayesha Butt
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Masooma Khawar
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Fazeeha Amjad
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| |
Collapse
|
20
|
TYK2 in Tumor Immunosurveillance. Cancers (Basel) 2020; 12:cancers12010150. [PMID: 31936322 PMCID: PMC7017180 DOI: 10.3390/cancers12010150] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/25/2019] [Indexed: 12/11/2022] Open
Abstract
We review the history of the tyrosine kinase 2 (TYK2) as the founding member of the Janus kinase (JAK) family and outline its structure-function relation. Gene-targeted mice and hereditary defects of TYK2 in men have established the biological and pathological functions of TYK2 in innate and adaptive immune responses to infection and cancer and in (auto-)inflammation. We describe the architecture of the main cytokine receptor families associated with TYK2, which activate signal transducers and activators of transcription (STATs). We summarize the cytokine receptor activities with well characterized dependency on TYK2, the types of cells that respond to cytokines and TYK2 signaling-induced cytokine production. TYK2 may drive beneficial or detrimental activities, which we explain based on the concepts of tumor immunoediting and the cancer-immunity cycle in the tumor microenvironment. Finally, we summarize current knowledge of TYK2 functions in mouse models of tumor surveillance. The biology and biochemistry of JAKs, TYK2-dependent cytokines and cytokine signaling in tumor surveillance are well covered in recent reviews and the oncogenic properties of TYK2 are reviewed in the recent Special Issue ‘Targeting STAT3 and STAT5 in Cancer’ of Cancers.
Collapse
|
21
|
Yang M, Tao B, Chen C, Jia W, Sun S, Zhang T, Wang X. Machine Learning Models Based on Molecular Fingerprints and an Extreme Gradient Boosting Method Lead to the Discovery of JAK2 Inhibitors. J Chem Inf Model 2019; 59:5002-5012. [DOI: 10.1021/acs.jcim.9b00798] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Minjian Yang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
- Joint Laboratory of Artificial Intelligence of the Institute of Materia Medica and Yuan Qi Zhi Yao, Beijing 100050, P.R. China
| | - Bingzhong Tao
- Joint Laboratory of Artificial Intelligence of the Institute of Materia Medica and Yuan Qi Zhi Yao, Beijing 100050, P.R. China
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Wenqiang Jia
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Shaolei Sun
- Joint Laboratory of Artificial Intelligence of the Institute of Materia Medica and Yuan Qi Zhi Yao, Beijing 100050, P.R. China
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
| | - Xiaojian Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P.R. China
- Joint Laboratory of Artificial Intelligence of the Institute of Materia Medica and Yuan Qi Zhi Yao, Beijing 100050, P.R. China
| |
Collapse
|
22
|
Bach J, Eastwood P, González J, Gómez E, Alonso JA, Fonquerna S, Lozoya E, Orellana A, Maldonado M, Calaf E, Albertí J, Pérez J, Andrés A, Prats N, Carreño C, Calama E, De Alba J, Calbet M, Miralpeix M, Ramis I. Identification of 2-Imidazopyridine and 2-Aminopyridone Purinones as Potent Pan-Janus Kinase (JAK) Inhibitors for the Inhaled Treatment of Respiratory Diseases. J Med Chem 2019; 62:9045-9060. [PMID: 31609613 DOI: 10.1021/acs.jmedchem.9b00533] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Janus kinases (JAKs) have a key role in regulating the expression and function of relevant inflammatory cytokines involved in asthma and chronic obstructive pulmonary disease. Herein are described the design, synthesis, and pharmacological evaluation of a series of novel purinone JAK inhibitors with profiles suitable for inhaled administration. Replacement of the imidazopyridine hinge binding motif present in the initial compounds of this series with a pyridone ring resulted in the mitigation of cell cytotoxicity. Further systematic structure-activity relationship (SAR) efforts driven by structural biology studies led to the discovery of pyridone 34, a potent pan-JAK inhibitor with good selectivity, long lung retention time, low oral bioavailability, and proven efficacy in the lipopolysaccharide-induced rat model of airway inflammation by the inhaled route.
Collapse
|
23
|
Identification of Chinese Herbal Compounds with Potential as JAK3 Inhibitors. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:4982062. [PMID: 31093295 PMCID: PMC6481137 DOI: 10.1155/2019/4982062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/07/2019] [Accepted: 03/17/2019] [Indexed: 01/30/2023]
Abstract
The Janus kinases (JAKs) consist of four similar tyrosine kinases and function as key hubs in the signaling pathways that are implicated in both innate and adaptive immunity. Among the four members, JAK3 is probably the more attractive target for treatment of inflammatory diseases because its inhibition demonstrates the greatest immunosuppression and most profound effect in the treatment of such disorders. Although many JAK3 inhibitors are already available, certain shortcomings have been identified, mostly acquired drug resistance or unwanted side effects. To discover and identify new promising lead candidates, in this study, the structure of JAK3 (3LXK) was obtained from the Protein Data Bank and used for simulation modeling and protein-ligand interaction analysis. The ~36,000 Chinese herbal compounds obtained from TCM Database@Taiwan were virtually screened by AutoDock Vina docking program and filtered with Lipinski's Rules and ADME/T virtual predictions. Because of high occurrence of fake hits during docking, we selected 12 phytochemicals which have demonstrated modulating JAKs expressions among the top 50 chemicals from docking results. To validate whether these compounds are able to directly mediate JAK3 kinase, we have investigated the inhibitory activity using enzymatic activity assays, western blot, and HEK 293 cell STAT5 transactivity assays. The molecular analysis included docking and molecular dynamics (MD) simulations in order to investigate structural conformations and to explore the key amino acids in the interaction between JAK3 kinase and its putative ligands. The results demonstrated that Cryptotanshinone, Icaritin, and Indirubin exhibited substantial inhibitory activity against JAK3 kinase in vitro. The results also provide binding models of the protein-ligand interaction, detailing the interacting amino acid residues at the active ATP-binding domains of JAK3 kinase. In conclusion, our work discovered 3 potential natural inhibitors of JAK3 kinase and could provide new possibilities and stimulate new insights for the treatment of JAK3-targeted diseases.
Collapse
|
24
|
Arora L, Kumar AP, Arfuso F, Chng WJ, Sethi G. The Role of Signal Transducer and Activator of Transcription 3 (STAT3) and Its Targeted Inhibition in Hematological Malignancies. Cancers (Basel) 2018; 10:cancers10090327. [PMID: 30217007 PMCID: PMC6162647 DOI: 10.3390/cancers10090327] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/22/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3), a member of the STAT protein family, can be phosphorylated by receptor-associated Janus kinases (JAKs) in response to stimulation by cytokines and growth factors. It forms homo- or heterodimers that can translocate to the cell nucleus where they act as transcription activators. Constitutive activation of STAT3 has been found to be associated with initiation and progression of various cancers. It can exert proliferative as well as anti-apoptotic effects. This review focuses on the role of STAT3 in pathogenesis i.e., proliferation, differentiation, migration, and apoptosis of hematological malignancies viz. leukemia, lymphoma and myeloma, and briefly highlights the potential therapeutic approaches developed against STAT3 activation pathway.
Collapse
Affiliation(s)
- Loukik Arora
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive, #11-01M, Singapore 117599, Singapore.
- Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia.
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore.
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive, #11-01M, Singapore 117599, Singapore.
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6009, Australia.
| |
Collapse
|
25
|
Siveen KS, Prabhu KS, Achkar IW, Kuttikrishnan S, Shyam S, Khan AQ, Merhi M, Dermime S, Uddin S. Role of Non Receptor Tyrosine Kinases in Hematological Malignances and its Targeting by Natural Products. Mol Cancer 2018; 17:31. [PMID: 29455667 PMCID: PMC5817858 DOI: 10.1186/s12943-018-0788-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Tyrosine kinases belong to a family of enzymes that mediate the movement of the phosphate group to tyrosine residues of target protein, thus transmitting signals from the cell surface to cytoplasmic proteins and the nucleus to regulate physiological processes. Non-receptor tyrosine kinases (NRTK) are a sub-group of tyrosine kinases, which can relay intracellular signals originating from extracellular receptor. NRTKs can regulate a huge array of cellular functions such as cell survival, division/propagation and adhesion, gene expression, immune response, etc. NRTKs exhibit considerable variability in their structural make up, having a shared kinase domain and commonly possessing many other domains such as SH2, SH3 which are protein-protein interacting domains. Recent studies show that NRTKs are mutated in several hematological malignancies, including lymphomas, leukemias and myelomas, leading to aberrant activation. It can be due to point mutations which are intragenic changes or by fusion of genes leading to chromosome translocation. Mutations that lead to constitutive kinase activity result in the formation of oncogenes, such as Abl, Fes, Src, etc. Therefore, specific kinase inhibitors have been sought after to target mutated kinases. A number of compounds have since been discovered, which have shown to inhibit the activity of NRTKs, which are remarkably well tolerated. This review covers the role of various NRTKs in the development of hematological cancers, including their deregulation, genetic alterations, aberrant activation and associated mutations. In addition, it also looks at the recent advances in the development of novel natural compounds that can target NRTKs and perhaps in combination with other forms of therapy can show great promise for the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Kodappully S. Siveen
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Kirti S. Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Iman W. Achkar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Sunitha Shyam
- Medical Research Center, Hamad Medical Corporation, Doha, State of Qatar
| | - Abdul Q. Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, State of Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, State of Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, State of Qatar
| |
Collapse
|
26
|
High Content Imaging Assays for IL-6-Induced STAT3 Pathway Activation in Head and Neck Cancer Cell Lines. Methods Mol Biol 2017; 1683:229-244. [PMID: 29082496 DOI: 10.1007/978-1-4939-7357-6_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
In the canonical STAT3 signaling pathway, IL-6 receptor engagement leads to the recruitment of latent STAT3 to the activated IL-6 complex and the associated Janus kinase (JAK) phosphorylates STAT3 at Y705. pSTAT3-Y705 dimers traffic into the nucleus and bind to specific DNA response elements in the promoters of target genes to regulate their transcription. However, IL-6 receptor activation induces the phosphorylation of both the Y705 and S727 residues of STAT3, and S727 phosphorylation is required to achieve maximal STAT3 transcriptional activity. STAT3 continuously shuttles between the nucleus and cytoplasm and maintains a prominent nuclear presence that is independent of Y705 phosphorylation. The constitutive nuclear entry of un-phosphorylated STAT3 (U-STAT3) drives expression of a second round of genes by a mechanism distinct from that used by pSTAT3-Y705 dimers. The abnormally elevated levels of U-STAT3 produced by the constitutive activation of pSTAT3-Y705 observed in many tumors drive the expression of an additional set of pSTAT3-independent genes that contribute to tumorigenesis. In this chapter, we describe the HCS assay methods to measure IL-6-induced STAT3 signaling pathway activation in head and neck tumor cell lines as revealed by the expression and subcellular distribution of pSTAT3-Y705, pSTAT3-S727, and U-STAT3. Only the larger dynamic range provided by the pSTAT3-Y705 antibody would be robust and reproducible enough for screening.
Collapse
|
27
|
Advances in the Development of Janus Kinase Inhibitors in Inflammatory Bowel Disease: Future Prospects. Drugs 2017; 77:1057-1068. [DOI: 10.1007/s40265-017-0755-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
28
|
Sen M, Johnston PA, Pollock NI, DeGrave K, Joyce SC, Freilino ML, Hua Y, Camarco DP, Close DA, Huryn DM, Wipf P, Grandis JR. Mechanism of action of selective inhibitors of IL-6 induced STAT3 pathway in head and neck cancer cell lines. J Chem Biol 2017; 10:129-141. [PMID: 28684999 DOI: 10.1007/s12154-017-0169-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/06/2017] [Indexed: 12/13/2022] Open
Abstract
Studies indicate that elevated interleukin-6 (IL-6) levels engage IL6Rα-gp130 receptor complexes to activate signal transducer and activator of transcription 3 (STAT3) that is hyperactivated in many cancers including head and neck squamous cell carcinoma (HNSCC). Our previous HCS campaign identified several hits that selectively blocked IL-6-induced STAT3 activation. This study describes our investigation of the mechanism(s) of action of three of the four chemical series that progressed to lead activities: a triazolothiadiazine (864669), amino alcohol (856350), and an oxazole-piperazine (4248543). We demonstrated that all three blocked IL-6-induced upregulation of the cyclin D1 and Bcl-XL STAT3 target genes. None of the compounds exhibited direct binding interactions with STAT3 in surface plasmon resonance (SPR) binding assays; neither did they inhibit the recruitment and binding of a phospho-tyrosine-gp130 peptide to STAT3 in a fluorescence polarization assay. Furthermore, they exhibited little or no inhibition in a panel of 83 cancer-associated in vitro kinase profiling assays, including lack of inhibition of IL-6-induced Janus kinase (JAK 1, 2, and 3) activation. Further, 864669 and 4248543 selectively inhibited IL-6-induced STAT3 activation but not that induced by oncostatin M (OSM). The compounds 864669 and 4248543 abrogated IL-6-induced phosphorylation of the gp130 signaling subunit (phospho-gp130Y905) of the IL-6-receptor complex in HNSCC cell lines which generate docking sites for the SH2 domains of STAT3. Our data indicate that 864669 and 4248543 block IL-6-induced STAT activation by interfering with the recruitment, assembly, or activation of the hexamer-activated IL-6/IL-6Rα/gp130 signaling complex that occurs after IL-6 binding to IL-6Rα subunits.
Collapse
Affiliation(s)
- Malabika Sen
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Paul A Johnston
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Netanya I Pollock
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Kara DeGrave
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Sonali C Joyce
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Maria L Freilino
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Yun Hua
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Daniel P Camarco
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - David A Close
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Donna M Huryn
- University of Pittsburgh Chemical Diversity Center, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Peter Wipf
- University of Pittsburgh Chemical Diversity Center, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Jennifer R Grandis
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94118 USA.,Clinical and Translational Science Institute, University of California, San Francisco, Box 0558, 550 16th Street, San Francisco, CA 94143 USA
| |
Collapse
|
29
|
Caspers NL, Han S, Rajamohan F, Hoth LR, Geoghegan KF, Subashi TA, Vazquez ML, Kaila N, Cronin CN, Johnson E, Kurumbail RG. Development of a high-throughput crystal structure-determination platform for JAK1 using a novel metal-chelator soaking system. Acta Crystallogr F Struct Biol Commun 2016; 72:840-845. [PMID: 27827355 PMCID: PMC5101585 DOI: 10.1107/s2053230x16016356] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/13/2016] [Indexed: 11/11/2022] Open
Abstract
Crystals of phosphorylated JAK1 kinase domain were initially generated in complex with nucleotide (ADP) and magnesium. The tightly bound Mg2+-ADP at the ATP-binding site proved recalcitrant to ligand displacement. Addition of a molar excess of EDTA helped to dislodge the divalent metal ion, promoting the release of ADP and allowing facile exchange with ATP-competitive small-molecule ligands. Many kinases require the presence of a stabilizing ligand in the ATP site for crystallization. This procedure could be useful for developing co-crystallization systems with an exchangeable ligand to enable structure-based drug design of other protein kinases.
Collapse
Affiliation(s)
- Nicole L. Caspers
- Structural Biology, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA
| | - Seungil Han
- Structural Biology, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA
| | - Francis Rajamohan
- Structural Biology, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA
| | - Lise R. Hoth
- Structural Biology, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA
| | | | - Timothy A. Subashi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA
| | - Michael L. Vazquez
- Inflammation Medicinal Chemistry, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Neelu Kaila
- Inflammation Medicinal Chemistry, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Ciarán N. Cronin
- Oncology Structural Biology, Pfizer Inc., 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Eric Johnson
- Oncology Structural Biology, Pfizer Inc., 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Ravi G. Kurumbail
- Structural Biology, Pfizer Inc., Eastern Point Road, Groton, CT 06340, USA
| |
Collapse
|
30
|
Johnston PA, Sen M, Hua Y, Camarco DP, Shun TY, Lazo JS, Wilson GM, Resnick LO, LaPorte MG, Wipf P, Huryn DM, Grandis JR. HCS campaign to identify selective inhibitors of IL-6-induced STAT3 pathway activation in head and neck cancer cell lines. Assay Drug Dev Technol 2016; 13:356-76. [PMID: 26317883 DOI: 10.1089/adt.2015.663] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Signal transducer and activator of transcription factor 3 (STAT3) is hyperactivated in head and neck squamous cell carcinomas (HNSCC). Cumulative evidence indicates that IL-6 production by HNSCC cells and/or stromal cells in the tumor microenvironment activates STAT3 and contributes to tumor progression and drug resistance. A library of 94,491 compounds from the Molecular Library Screening Center Network (MLSCN) was screened for the ability to inhibit interleukin-6 (IL-6)-induced pSTAT3 activation. For contractual reasons, the primary high-content screening (HCS) campaign was conducted over several months in 3 distinct phases; 1,068 (1.1%) primary HCS actives remained after cytotoxic or fluorescent outliers were eliminated. One thousand one hundred eighty-seven compounds were cherry-picked for confirmation; actives identified in the primary HCS and compounds selected by a structural similarity search of the remaining MLSCN library using hits identified in phases I and II of the screen. Actives were confirmed in pSTAT3 IC50 assays, and an IFNγ-induced pSTAT1 activation assay was used to prioritize selective inhibitors of STAT3 activation that would not inhibit STAT1 tumor suppressor functions. Two hundred three concentration-dependent inhibitors of IL-6-induced pSTAT3 activation were identified and 89 of these also produced IC50s against IFN-γ-induced pSTAT1 activation. Forty-nine compounds met our hit criteria: they reproducibly inhibited IL-6-induced pSTAT3 activation by ≥70% at 20 μM; their pSTAT3 activation IC50s were ≤25 μM; they were ≥2-fold selective for pSTAT3 inhibition over pSTAT1 inhibition; a cross target query of PubChem indicated that they were not biologically promiscuous; and they were ≥90% pure. Twenty-six chemically tractable hits that passed filters for nuisance compounds and had acceptable drug-like and ADME-Tox properties by computational evaluation were purchased for characterization. The hit structures were distributed among 5 clusters and 8 singletons. Twenty-four compounds inhibited IL-6-induced pSTAT3 activation with IC50s ≤20 μM and 13 were ≥3-fold selective versus inhibition of pSTAT1 activation. Eighteen hits inhibited the growth of HNSCC cell lines with average IC50s ≤ 20 μM. Four chemical series were progressed into lead optimization: the guanidinoquinazolines, the triazolothiadiazines, the amino alcohols, and an oxazole-piperazine singleton.
Collapse
Affiliation(s)
- Paul A Johnston
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania.,2 University of Pittsburgh Cancer Institute , Pittsburgh, Pennsylvania.,3 Pittsburgh Specialized Application Center, University of Pittsburgh Drug Discovery Institute , Pittsburgh, Pennsylvania
| | - Malabika Sen
- 4 Department of Otolaryngology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Yun Hua
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Daniel P Camarco
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Tong Ying Shun
- 3 Pittsburgh Specialized Application Center, University of Pittsburgh Drug Discovery Institute , Pittsburgh, Pennsylvania
| | - John S Lazo
- 5 Departments of Pharmacology and Chemistry, University of Virginia , Charlottesville, Virginia
| | - Gabriela Mustata Wilson
- 6 University of Pittsburgh Chemical Diversity Center , Pittsburgh, Pennsylvania.,7 Department of Health Services and Health Administration, College of Nursing and Health Professions, University of Southern Indiana , Evansville, Indiana
| | - Lynn O Resnick
- 6 University of Pittsburgh Chemical Diversity Center , Pittsburgh, Pennsylvania
| | - Matthew G LaPorte
- 6 University of Pittsburgh Chemical Diversity Center , Pittsburgh, Pennsylvania
| | - Peter Wipf
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania.,2 University of Pittsburgh Cancer Institute , Pittsburgh, Pennsylvania.,6 University of Pittsburgh Chemical Diversity Center , Pittsburgh, Pennsylvania.,8 Department of Chemistry, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Donna M Huryn
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania.,6 University of Pittsburgh Chemical Diversity Center , Pittsburgh, Pennsylvania
| | - Jennifer R Grandis
- 9 Clinical and Translational Science Institute, Otolaryngology - Head and Neck Surgery, University of California , San Francisco, California
| |
Collapse
|
31
|
Roskoski R. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacol Res 2016; 111:784-803. [PMID: 27473820 DOI: 10.1016/j.phrs.2016.07.038] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 02/06/2023]
Abstract
The Janus kinase (JAK) family of non-receptor protein-tyrosine kinases consists of JAK1, JAK2, JAK3, and TYK2 (tyrosine kinase-2). Each of these proteins contains a JAK homology pseudokinase (JH2) domain that regulates the adjacent protein kinase domain (JH1). JAK1/2 and TYK2 are ubiquitously expressed whereas JAK3 is found predominantly in hematopoietic cells. The Janus kinase family is regulated by numerous cytokines including interleukins, interferons, and hormones such as erythropoietin, thrombopoietin, and growth hormone. Ligand binding to cytokine and hormone receptors leads to the activation of associated Janus kinases, which then mediate the phosphorylation of the receptors. The SH2 domain of STATs (signal transducers and activators of transcription) binds to the receptor phosphotyrosines thereby promoting STAT phosphorylation by the Janus kinases and consequent activation. STAT dimers are translocated to the nucleus where they participate in the regulation of the expression of thousands of proteins. JAK-STAT dysregulation results in autoimmune disorders such as rheumatoid arthritis, ulcerative colitis, and Crohn disease. JAK-STAT dysregulation also plays a role in the pathogenesis of myelofibrosis, polycythemia vera, and other myeloproliferative illnesses. An activating JAK2 V617F mutation occurs in 95% of people with polycythemia vera and in a lower percentage of people with other neoplasms. JAK1/3 signaling participates in the pathogenesis of inflammatory afflictions while JAK1/2 signaling participates in the development of several malignancies including leukemias and lymphomas as well as myeloproliferative neoplasms. Tofacitinib is a pan-JAK inhibitor that is approved by the FDA for the treatment of rheumatoid arthritis and ruxolitinib is a JAK1/2 inhibitor that is approved for the treatment of polycythemia vera and myelofibrosis.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 116, Box 19, Horse Shoe, NC 28742-8814, United States.
| |
Collapse
|
32
|
Zimmers TA, Fishel ML, Bonetto A. STAT3 in the systemic inflammation of cancer cachexia. Semin Cell Dev Biol 2016; 54:28-41. [PMID: 26860754 PMCID: PMC4867234 DOI: 10.1016/j.semcdb.2016.02.009] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/04/2016] [Indexed: 02/07/2023]
Abstract
Weight loss is diagnostic of cachexia, a debilitating syndrome contributing mightily to morbidity and mortality in cancer. Most research has probed mechanisms leading to muscle atrophy and adipose wasting in cachexia; however cachexia is a truly systemic phenomenon. Presence of the tumor elicits an inflammatory response and profound metabolic derangements involving not only muscle and fat, but also the hypothalamus, liver, heart, blood, spleen and likely other organs. This global response is orchestrated in part through circulating cytokines that rise in conditions of cachexia. Exogenous Interleukin-6 (IL6) and related cytokines can induce most cachexia symptomatology, including muscle and fat wasting, the acute phase response and anemia, while IL-6 inhibition reduces muscle loss in cancer. Although mechanistic studies are ongoing, certain of these cachexia phenotypes have been causally linked to the cytokine-activated transcription factor, STAT3, including skeletal muscle wasting, cardiac dysfunction and hypothalamic inflammation. Correlative studies implicate STAT3 in fat wasting and the acute phase response in cancer cachexia. Parallel data in non-cancer models and disease states suggest both pathological and protective functions for STAT3 in other organs during cachexia. STAT3 also contributes to cancer cachexia through enhancing tumorigenesis, metastasis and immune suppression, particularly in tumors associated with high prevalence of cachexia. This review examines the evidence linking STAT3 to multi-organ manifestations of cachexia and the potential and perils for targeting STAT3 to reduce cachexia and prolong survival in cancer patients.
Collapse
Affiliation(s)
- Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| | - Melissa L Fishel
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States; IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| |
Collapse
|
33
|
Wang T, Liu X, Hao M, Qiao J, Ju C, Xue L, Zhang C. Design, synthesis and evaluation of pyrrolo[2,3- d ]pyrimidine-phenylamide hybrids as potent Janus kinase 2 inhibitors. Bioorg Med Chem Lett 2016; 26:2936-2941. [DOI: 10.1016/j.bmcl.2016.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/07/2016] [Accepted: 04/12/2016] [Indexed: 12/31/2022]
|
34
|
Zimmermann K, Sang X, Mastalerz HA, Johnson WL, Zhang G, Liu Q, Batt D, Lombardo LJ, Vyas D, Trainor GL, Tokarski JS, Lorenzi MV, You D, Gottardis MM, Lippy J, Khan J, Sack JS, Purandare AV. 9H-Carbazole-1-carboxamides as potent and selective JAK2 inhibitors. Bioorg Med Chem Lett 2015; 25:2809-12. [PMID: 25987372 DOI: 10.1016/j.bmcl.2015.04.101] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 04/28/2015] [Accepted: 04/30/2015] [Indexed: 02/07/2023]
Abstract
The discovery, synthesis, and characterization of 9H-carbazole-1-carboxamides as potent and selective ATP-competitive inhibitors of Janus kinase 2 (JAK2) are discussed. Optimization for JAK family selectivity led to compounds 14 and 21, with greater than 45-fold selectivity for JAK2 over all other members of the JAK kinase family.
Collapse
Affiliation(s)
- Kurt Zimmermann
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA.
| | - Xiaopeng Sang
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - Harold A Mastalerz
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - Walter L Johnson
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - Guifen Zhang
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - Qingjie Liu
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Douglas Batt
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Louis J Lombardo
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Dinesh Vyas
- Bristol-Myers Squibb Co., 5 Research Parkway, Wallingford, CT 06492-1951, USA
| | - George L Trainor
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - John S Tokarski
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Matthew V Lorenzi
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Dan You
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Marco M Gottardis
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Jonathan Lippy
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Javed Khan
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - John S Sack
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| | - Ashok V Purandare
- Bristol-Myers Squibb Co., Province Line Rd., Princeton, NJ 08540-4000, USA
| |
Collapse
|
35
|
Carbone CJ, Fuchs SY. Eliminative signaling by Janus kinases: role in the downregulation of associated receptors. J Cell Biochem 2014; 115:8-16. [PMID: 23959845 DOI: 10.1002/jcb.24647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 07/31/2013] [Indexed: 12/11/2022]
Abstract
Activation of cytokine receptor-associated Janus kinases (JAKs) mediates most, if not all, of the cellular responses to peptide hormones and cytokines. Consequently, JAKs play a paramount role in homeostasis and immunity. Members of this family of tyrosine kinases control the cytokine/hormone-induced alterations in cell gene expression program. This function is largely mediated through an ability to signal toward activation of the signal transducer and activator of transcription proteins (STAT), as well as toward some other pathways. Importantly, JAKs are also instrumental in tightly controlling the expression of associated cytokine and hormone receptors, and, accordingly, in regulating the cell sensitivity to these cytokines and hormones. This review highlights the enzymatic and non-enzymatic mechanisms of this regulation and discusses the importance of the ambidextrous nature of JAK as a key signaling node that integrates the combining functions of forward signaling and eliminative signaling. Attention to the latter aspect of JAK function may contribute to emancipating our approaches to the pharmacological modulation of JAKs.
Collapse
Affiliation(s)
- Christopher J Carbone
- Department of Animal Biology and Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104
| | | |
Collapse
|
36
|
Kim MK, Bae O, Chong Y. Design, Synthesis, and Molecular Docking Study of Flavonol Derivatives as Selective JAK1 Inhibitors. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.8.2581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
37
|
Siveen KS, Sikka S, Surana R, Dai X, Zhang J, Kumar AP, Tan BKH, Sethi G, Bishayee A. Targeting the STAT3 signaling pathway in cancer: role of synthetic and natural inhibitors. Biochim Biophys Acta Rev Cancer 2014; 1845:136-54. [PMID: 24388873 DOI: 10.1016/j.bbcan.2013.12.005] [Citation(s) in RCA: 374] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 12/24/2013] [Accepted: 12/27/2013] [Indexed: 12/25/2022]
Abstract
Signal transducers and activators of transcription (STATs) comprise a family of cytoplasmic transcription factors that mediate intracellular signaling that is usually generated at cell surface receptors and thereby transmit it to the nucleus. Numerous studies have demonstrated constitutive activation of STAT3 in a wide variety of human tumors, including hematological malignancies (leukemias, lymphomas, and multiple myeloma) as well as diverse solid tumors (such as head and neck, breast, lung, gastric, hepatocellular, colorectal and prostate cancers). There is strong evidence to suggest that aberrant STAT3 signaling promotes initiation and progression of human cancers by either inhibiting apoptosis or inducing cell proliferation, angiogenesis, invasion, and metastasis. Suppression of STAT3 activation results in the induction of apoptosis in tumor cells, and accordingly its pharmacological modulation by tyrosine kinase inhibitors, antisense oligonucleotides, decoy nucleotides, dominant negative proteins, RNA interference and chemopreventive agents have been employed to suppress the proliferation of various human cancer cells in culture and tumorigenicity in vivo. However, the identification and development of novel drugs that can target deregulated STAT3 activation effectively remains an important scientific and clinical challenge. This review presents the evidence for critical roles of STAT3 in oncogenesis and discusses the potential for development of novel cancer therapies based on mechanistic understanding of STAT3 signaling cascade.
Collapse
Affiliation(s)
| | - Sakshi Sikka
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore
| | - Rohit Surana
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore
| | - Xiaoyun Dai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jingwen Zhang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore; School of Biomedical Sciences, Faculty of Health Sciences, Curtin University, Western Australia, Australia; Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Benny K H Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, School of Pharmacy, American University of Health Sciences, Signal Hill, CA, USA.
| |
Collapse
|
38
|
Johnston PA, Sen M, Hua Y, Camarco D, Shun TY, Lazo JS, Grandis JR. High-content pSTAT3/1 imaging assays to screen for selective inhibitors of STAT3 pathway activation in head and neck cancer cell lines. Assay Drug Dev Technol 2014; 12:55-79. [PMID: 24127660 PMCID: PMC3934522 DOI: 10.1089/adt.2013.524] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) is hyperactivated in most cancers and represents a plausible therapeutic target. In the absence of STAT3-selective small-molecule inhibitors, we sought to develop pSTAT3/1 high-content imaging (HCS) assays to screen for selective inhibitors of STAT3 pathway activation in head and neck squamous cell carcinomas (HNSCC) tumor cell lines. Based on the expression of the interleukin-6 (IL-6)Rα and gp130 subunits of the IL-6 receptor complex and STAT3, we selected the Cal33 HNSCC cell line as our model. After developing image acquisition and analysis procedures, we rigorously investigated the cytokine activation responses to optimize the dynamic ranges of both assays and demonstrated that the pan-Janus kinase inhibitor pyridone 6 nonselectively inhibited pSTAT3 and pSTAT1 activation with 50% inhibition concentrations of 7.19 ± 4.08 and 16.38 ± 8.45 nM, respectively. The optimized pSTAT3 HCS assay performed very well in a pilot screen of 1,726 compounds from the Library of Pharmacologically Active Compounds and the National Institutes of Health clinical collection sets, and we identified 51 inhibitors of IL-6-induced pSTAT3 activation. However, only three of the primary HCS actives selectively inhibited STAT3 compared with STAT1. Our follow-up studies indicated that the nonselective inhibition of cytokine induced pSTAT3 and pSTAT1 activation by G-alpha stimulatory subunit-coupled G-protein-coupled receptor agonists, and forskolin was likely due to cyclic adenosine monophosphate-mediated up-regulation of suppressors of cytokine signaling 3. Azelastine, an H1 receptor antagonist approved for the treatment of seasonal allergic rhinitis, nonallergic vasomotor rhinitis, and ocular conjunctivitis, was subsequently confirmed as a selective inhibitor of IL-6-induced pSTAT3 activation that also reduced the growth of HNSCC cell lines. These data illustrate the power of a chemical biology approach to lead generation that utilizes fully developed and optimized HCS assays as phenotypic screens to interrogate specific signaling pathways.
Collapse
Affiliation(s)
- Paul A. Johnston
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Malabika Sen
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yun Hua
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel Camarco
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tong Ying Shun
- Department of Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John S. Lazo
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
- Department of Chemistry, University of Virginia, Charlottesville, Virginia
| | - Jennifer R. Grandis
- Department of University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
39
|
Leoni A, Locatelli A, Morigi R, Rambaldi M. Novel thiazole derivatives: a patent review (2008 – 2012; Part 1). Expert Opin Ther Pat 2013; 24:201-16. [DOI: 10.1517/13543776.2014.858121] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
40
|
Differential selectivity of JAK2 inhibitors in enzymatic and cellular settings. Exp Hematol 2013; 41:491-500. [DOI: 10.1016/j.exphem.2013.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 01/08/2013] [Accepted: 01/16/2013] [Indexed: 01/09/2023]
|
41
|
Abstract
JAK kinases are critical mediators in development, differentiation, and homeostasis and accordingly, have become well-validated targets for drug discovery efforts. In recent years, the integration of X-ray crystallography in kinase-focused drug discovery programs has provided a powerful rationale for chemical modification by allowing a unique glimpse of a bound inhibitor to its target. Such structural information has not only led to an improved understanding of the key drivers of potency and specificity of several JAK-specific compounds but has greatly facilitated and accelerated the design of compounds with improved pharmacokinetic properties.JAK kinases are traditionally difficult candidates to express in significant quantities, generally requiring eukaryotic expression systems, protein engineering, mutations to yield soluble, homogeneous samples suitable for crystallization studies. Here we review the key methods utilized to express, purify, and crystallize the JAK kinases and provide a detail description of the methods that we have developed to express, purify, and crystallize recombinant JAK1 and JAK2 proteins in the presence of small molecule inhibitors.
Collapse
|
42
|
Kim MK, Chong Y. Towards Selective Inhibitors of Janus Kinase 3: Identification of a Novel Structural Variation between Janus Kinases 2 and 3. B KOREAN CHEM SOC 2012. [DOI: 10.5012/bkcs.2012.33.12.4207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
Zhou C, Baltz JM. JAK2 mediates the acute response to decreased cell volume in mouse preimplantation embryos by activating NHE1. J Cell Physiol 2012; 228:428-38. [DOI: 10.1002/jcp.24147] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
44
|
Nicolas CS, Peineau S, Amici M, Csaba Z, Fafouri A, Javalet C, Collett VJ, Hildebrandt L, Seaton G, Choi SL, Sim SE, Bradley C, Lee K, Zhuo M, Kaang BK, Gressens P, Dournaud P, Fitzjohn SM, Bortolotto ZA, Cho K, Collingridge GL. The Jak/STAT pathway is involved in synaptic plasticity. Neuron 2012; 73:374-90. [PMID: 22284190 PMCID: PMC3268861 DOI: 10.1016/j.neuron.2011.11.024] [Citation(s) in RCA: 190] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2011] [Indexed: 12/15/2022]
Abstract
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway is involved in many cellular processes, including cell growth and differentiation, immune functions and cancer. It is activated by various cytokines, growth factors, and protein tyrosine kinases (PTKs) and regulates the transcription of many genes. Of the four JAK isoforms and seven STAT isoforms known, JAK2 and STAT3 are highly expressed in the brain where they are present in the postsynaptic density (PSD). Here, we demonstrate a new neuronal function for the JAK/STAT pathway. Using a variety of complementary approaches, we show that the JAK/STAT pathway plays an essential role in the induction of NMDA-receptor dependent long-term depression (NMDAR-LTD) in the hippocampus. Therefore, in addition to established roles in cytokine signaling, the JAK/STAT pathway is involved in synaptic plasticity in the brain.
Collapse
Affiliation(s)
- Céline S Nicolas
- MRC Centre for Synaptic Plasticity, School of Physiology and Pharmacology, Medical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Prchal-Murphy M, Semper C, Lassnig C, Wallner B, Gausterer C, Teppner-Klymiuk I, Kobolak J, Müller S, Kolbe T, Karaghiosoff M, Dinnyés A, Rülicke T, Leitner NR, Strobl B, Müller M. TYK2 kinase activity is required for functional type I interferon responses in vivo. PLoS One 2012; 7:e39141. [PMID: 22723949 PMCID: PMC3377589 DOI: 10.1371/journal.pone.0039141] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 05/20/2012] [Indexed: 01/25/2023] Open
Abstract
Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family and is involved in cytokine signalling. In vitro analyses suggest that TYK2 also has kinase-independent, i.e., non-canonical, functions. We have generated gene-targeted mice harbouring a mutation in the ATP-binding pocket of the kinase domain. The Tyk2 kinase-inactive (Tyk2K923E) mice are viable and show no gross abnormalities. We show that kinase-active TYK2 is required for full-fledged type I interferon- (IFN) induced activation of the transcription factors STAT1-4 and for the in vivo antiviral defence against viruses primarily controlled through type I IFN actions. In addition, TYK2 kinase activity was found to be required for the protein’s stability. An inhibitory function was only observed upon over-expression of TYK2K923Ein vitro. Tyk2K923E mice represent the first model for studying the kinase-independent function of a JAK in vivo and for assessing the consequences of side effects of JAK inhibitors.
Collapse
Affiliation(s)
- Michaela Prchal-Murphy
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Christian Semper
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Caroline Lassnig
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
- Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
| | - Barbara Wallner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Christian Gausterer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | | | - Julianna Kobolak
- Genetic Reprogramming Group Agricultural Biotechnology Center, Gödöllö, Hungary
| | - Simone Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
- Department for Agrobiotechnology IFA Tulln, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Marina Karaghiosoff
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Andras Dinnyés
- Genetic Reprogramming Group Agricultural Biotechnology Center, Gödöllö, Hungary
- Molecular Animal Biotechnology Laboratory, Szent Istvan University, Gödöllö, Hungary
- BioTalentum Ltd., Gödöllö, Hungary
| | - Thomas Rülicke
- Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
- Institute of Laboratory Animal Science, University of Veterinary Medicine, Vienna, Austria
| | - Nicole R. Leitner
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
- Biomodels Austria, University of Veterinary Medicine, Vienna, Austria
- * E-mail:
| |
Collapse
|
46
|
Zhang H, Photiou A, Grothey A, Stebbing J, Giamas G. The role of pseudokinases in cancer. Cell Signal 2012; 24:1173-84. [PMID: 22330072 DOI: 10.1016/j.cellsig.2012.01.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 01/27/2012] [Indexed: 01/12/2023]
Abstract
Kinases play a critical role in regulating many cellular functions including development, differentiation and proliferation. To date, over 518 proteins with kinase activity, comprising ~2-3% of total cellular proteins, have been identified from within the human kinome. Interestingly, approximately 10% of kinases are categorised as pseudokinases since they lack one or more conserved catalytic residues within their kinase domain and were originally thought to have no enzymatic activity. Recently, there has been strong evidence to suggest that some pseudokinsases can not only function as scaffold proteins, but may also possess kinase activity leading to modulation of cell signalling pathways. Altered activity of these pseudokinases can result in impaired cellular function, particularly in malignancies. In this review we are discussing recent evidence that apart from a scaffolding role, pseudokinases also orchestrate cellular processes as active kinases per se in signalling pathways of malignant cells.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Cancer and Surgery, Imperial College, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | | | | | | | | |
Collapse
|
47
|
Black SM, Benson BA, Idossa D, Vercellotti GM, Dalmasso AP. Protection of porcine endothelial cells against apoptosis with interleukin-4. Xenotransplantation 2012; 18:343-54. [PMID: 22168141 DOI: 10.1111/j.1399-3089.2011.00678.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Apoptosis is crucial for tissue development and homeostasis, and insufficient apoptosis is pivotal in cancer pathogenesis. Apoptosis may also be important in tissue injury and in this case, it is of interest to induce protection against apoptosis. In organ transplantation, apoptosis has been implicated in acute vascular rejection (AVR); in xenotransplantation, the inducers of apoptosis of relevance in AVR, such as tumor necrosis factor-α (TNF-α), also cause endothelial cell (EC) activation. We have previously shown that interleukin (IL)-4 and IL-13 induced protection in porcine ECs against activation and apoptosis triggered by TNF-α. Now we define signaling processes activated by IL-4 in porcine ECs and mechanisms required for IL-4-induced protection against apoptosis. METHODS Porcine aortic ECs were used as primary cultures or as virus-induced immortalized cells derived from galactosyl transferase-deficient (Gal(-/-) ) or wild-type pigs. ECs were stimulated with porcine IL-4, either extrinsically or transduced with recombinant adenovirus (adeno) IL-4, and analyzed using immunoblotting. Apoptosis was induced with TNF-α plus cycloheximide and assessed using neutral red uptake or flow cytometry. The role of various signaling proteins in IL-4-induced protection was established using pharmacologic inhibitors and siRNA downregulation of protein expression. RESULTS IL-4 induced similar degrees of phosphorylation of STAT6 in all 3 types of ECs, and STAT6 was phosphorylated through Jak3. IL-4 induced phosphorylation of Bad through Jak3. Stimulation of ECs with IL-4 caused protection of ECs against apoptosis with an absolute requirement of Jak3/STAT6 activation and major participation of mammalian target of rapamycin complex 2 (mTORC2), Akt, and extracellular signal-regulated kinase 1/2. IL-4 caused no increase in EC levels of protective proteins hemoxygenase-1, inhibitor of apoptosis protein, heat shock protein 70, Bcl-2, and Bcl-xL. ECs transduced with adenoIL-4 exhibited strong and durable protection from apoptosis. Gal(-/-) ECs were as susceptible to induction of protection with IL-4 as wild-type ECs. CONCLUSIONS IL-4 induces activation of Jak3/STAT6 and phosphorylation of Bad in porcine ECs, ultimately resulting in effective protection of the ECs from apoptosis. Delineation of downstream signals activated by IL-4 that are required for induction of protection suggests possible sites of intervention to design effective therapeutic agents. This is of interest because substances such as IL-4 have pleiotropic effects and cannot be used directly due to potential deleterious effects. Inducing resistance to apoptosis in porcine vascular endothelium may be important to facilitate xenograft survival and accommodation.
Collapse
Affiliation(s)
- Sylvester M Black
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
48
|
Zouein FA, Duhé RJ, Booz GW. JAKs go nuclear: emerging role of nuclear JAK1 and JAK2 in gene expression and cell growth. Growth Factors 2011; 29:245-52. [PMID: 21892841 PMCID: PMC3595105 DOI: 10.3109/08977194.2011.614949] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The four Janus kinases (JAKs) comprise a family of intracellular, nonreceptor tyrosine kinases that first gained attention as signaling mediators of the type I and type II cytokine receptors. Subsequently, the JAKs were found to be involved in signaling downstream of the insulin receptor, a number of receptor tyrosine kinases, and certain G-protein coupled receptors. Although a number of cytoplasmic targets for the JAKs have been identified, their predominant action was found to be the phosphorylation and activation of the signal transducers and activators of transcription (STAT) factors. Through the STATs, the JAKs activate gene expression linked to cellular stress, proliferation, and differentiation. The JAKs are especially important in hematopoiesis, inflammation, and immunity, and aberrant JAK activity has been implicated in a number of disorders including rheumatoid arthritis, psoriasis, polycythemia vera, and myeloproliferative diseases. Although once thought to reside strictly in the cytoplasm, recent evidence shows that JAK1 and JAK2 are present in the nucleus of certain cells often under conditions associated with high rates of cell growth. Nuclear JAKs have now been shown to affect gene expression by activating other transcription factors besides the STATs and exerting epigenetic actions, for example, by phosphorylating histone H3. The latter action derepresses global gene expression and has been implicated in leukemogenesis. Nuclear JAKs may have a role as well in stem cell biology. Here we describe recent developments in understanding the noncanonical nuclear actions of JAK1 and JAK2.
Collapse
Affiliation(s)
- Fouad A. Zouein
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, Mississippi, USA
- The Center for Excellence in Cardiovascular-Renal Research, The University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Roy J. Duhé
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, Mississippi, USA
- University of Mississippi Cancer Institute, The University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, Mississippi, USA
- The Center for Excellence in Cardiovascular-Renal Research, The University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
49
|
Qian CJ, Yao J, Si JM. Nuclear JAK2: form and function in cancer. Anat Rec (Hoboken) 2011; 294:1446-59. [PMID: 21809458 DOI: 10.1002/ar.21443] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 05/19/2011] [Indexed: 12/23/2022]
Abstract
The conventional view of Janus kinase 2 (JAK2) is a nonreceptor tyrosine kinase which transmits information to the nucleus via the signal transducer and activator of transcriptions (STATs) without leaving the cytoplasm. However, accumulating data suggest that JAK2 may signal by exporting from cytoplasm to nucleus, where it guides the transcriptional machinery independent of STATs protein. Recent studies demonstrated that JAK2 is a crucial component of signaling pathways operating in the nucleus. Especially the latest landmark discovery confirmed that JAK2 goes into the nucleus and directly interacts with nucleoproteins, such as histone H3 at tyrosine 41 (H3Y41), nuclear factor 1-C2 (NF1-C2) and SWI/SNF-related helicases/ATPases (RUSH)-1α, indicating that JAK2 has a fresh nuclear function. Nuclear JAK2 is linked to a variety of cellular functions, such as cell cycle progression, apoptosis and genetic instability. The balance between these functions is an essential factor in determining whether a cell remains benign or becomes malignant. The aim of this review is intended to summarize the state of our knowledge on nuclear localization of JAK2 and nuclear JAK2 pathways, and to highlight the emerging roles for nuclear JAK2 in carcinogenesis.
Collapse
Affiliation(s)
- Cui-Juan Qian
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | |
Collapse
|
50
|
Johnston PA, Grandis JR. STAT3 signaling: anticancer strategies and challenges. Mol Interv 2011; 11:18-26. [PMID: 21441118 DOI: 10.1124/mi.11.1.4] [Citation(s) in RCA: 332] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Multiple lines of evidence place STAT3 at a central node in the development, progression, and maintenance of many human tumors, and STAT3 has been validated as an anti-cancer target in several contexts. STAT3 modulates the transcription of a variety of genes involved in the regulation of critical functions, including cell differentiation, proliferation, apoptosis, angiogenesis, metastasis, and immune responses. For many cancers, elevated levels of activated STAT3 have been associated with a poor prognosis. We review approaches that have been pursued to target STAT3, and we highlight some of the promises and challenges associated with developing an anticancer drug that might therapeutically inhibit the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Paul A Johnston
- School of Pharmaceutical Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|